Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Endocrine ; 81(3): 492-502, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37306934

RESUMEN

PURPOSE: In type 2 Diabetes, ß-cell failure is caused by loss of cell mass, mostly by apoptosis, but also by simple dysfunction (dedifferentiation, decline of glucose-stimulated insulin secretion). Apoptosis and dysfunction are caused, at least in part, by glucotoxicity, in which increased flux of glucose in the hexosamine biosynthetic pathway plays a role. In this study, we sought to clarify whether increased hexosamine biosynthetic pathway flux affects another important aspect of ß-cell physiology, that is ß-cell-ß-cell homotypic interactions. METHODS: We used INS-1E cells and murine islets. The expression and cellular distribution of E-cadherin and ß-catenin was evaluated by immunofluorescence, immunohistochemistry and western blot. Cell-cell adhesion was examined by the hanging-drop aggregation assay, islet architecture by isolation and microscopic observation. RESULTS: E-cadherin expression was not changed by increased hexosamine biosynthetic pathway flux, however, there was a decrease of cell surface, and an increase in intracellular E-cadherin. Moreover, intracellular E-cadherin delocalized, at least in part, from the Golgi complex to the endoplasmic reticulum. Beta-catenin was found to parallel the E-cadherin redistribution, showing a dislocation from the plasmamembrane to the cytosol. These changes had as a phenotypic consequence a decreased ability of INS-1E to aggregate. Finally, in ex vivo experiments, glucosamine was able to alter islet structure and to decrease surface abundandance of E-cadherin and ß-catenin. CONCLUSION: Increased hexosamine biosynthetic pathway flux alters E-cadherin cellular localization both in INS-1E cells and murine islets and affects cell-cell adhesion and islet morphology. These changes are likely caused by alterations of E-cadherin function, highlighting a new potential target to counteract the consequences of glucotoxicity on ß-cells.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Ratones , Animales , Insulina/metabolismo , beta Catenina/metabolismo , Hexosaminas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Adhesión Celular , Vías Biosintéticas , Células Secretoras de Insulina/metabolismo , Glucosa/metabolismo , Cadherinas/metabolismo , Islotes Pancreáticos/metabolismo
2.
Int J Mol Sci ; 23(21)2022 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-36362390

RESUMEN

The primary functional units of the thyroid gland are follicles of various sizes comprised of a monolayer of epithelial cells (thyrocytes) surrounding an apical extracellular cavity known as the follicle lumen. In the normal thyroid gland, the follicle lumen is filled with secreted protein (referred to as colloid), comprised nearly exclusively of thyroglobulin with a half-life ranging from days to weeks. At the cellular boundary of the follicle lumen, secreted thyroglobulin becomes iodinated, resulting from the coordinated activities of enzymes localized to the thyrocyte apical plasma membrane. Thyroglobulin appearance in evolution is essentially synchronous with the appearance of the follicular architecture of the vertebrate thyroid gland. Thyroglobulin is the most highly expressed thyroid gene and represents the most abundantly expressed thyroid protein. Wildtype thyroglobulin protein is a large and complex glycoprotein that folds in the endoplasmic reticulum, leading to homodimerization and export via the classical secretory pathway to the follicle lumen. However, of the hundreds of human thyroglobulin genetic variants, most exhibit increased susceptibility to misfolding with defective export from the endoplasmic reticulum, triggering hypothyroidism as well as thyroidal endoplasmic reticulum stress. The human disease of hypothyroidism with defective thyroglobulin (either homozygous, or compound heterozygous) can be experimentally modeled in thyrocyte cell culture, or in whole animals, such as mice that are readily amenable to genetic manipulation. From a combination of approaches, it can be demonstrated that in the setting of thyroglobulin misfolding, thyrocytes under chronic continuous ER stress exhibit increased susceptibility to cell death, with interesting cell biological and pathophysiological consequences.


Asunto(s)
Hipotiroidismo , Células Epiteliales Tiroideas , Ratones , Humanos , Animales , Tiroglobulina/metabolismo , Hipotiroidismo/metabolismo , Células Epiteliales Tiroideas/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas/metabolismo
3.
Front Endocrinol (Lausanne) ; 11: 588685, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240221

RESUMEN

The endoplasmic reticulum stress and the unfolded protein response are triggered following an imbalance between protein load and protein folding. Until recently, two possible outcomes of the unfolded protein response have been considered: life or death. We sought to substantiate a third alternative, dedifferentiation, mesenchymal shift, and activation of the antioxidant response by using typical endocrine cells, i.e. thyroid cells. The thyroid is a unique system both of endoplasmic reticulum stress (a single protein, thyroglobulin represents the majority of proteins synthesized in the endoplasmic reticulum by the thyrocyte) and of polarized epithelium (the single layer of thyrocytes delimiting the follicle). Following endoplasmic reticulum stress, in thyroid cells the folding of thyroglobulin was disrupted. The mRNAs of unfolded protein response were induced or spliced (X-box binding protein-1). Differentiation was inhibited: mRNA levels of thyroid specific genes, and of thyroid transcription factors were dramatically downregulated, at least in part, transcriptionally. The dedifferentiating response was accompanied by an upregulation of mRNAs of antioxidant genes. Moreover, cadherin-1, and the thyroid (and kidney)-specific cadherin-16 mRNAs were downregulated, vimentin, and SNAI1 mRNAs were upregulated. In addition, loss of cortical actin and stress fibers formation were observed. Together, these data indicate that ER stress in thyroid cells induces dedifferentiation, loss of epithelial organization, shift towards a mesenchymal phenotype, and activation of the antioxidant response, highlighting, at the same time, a new and wide strategy to achieve survival following ER stress, and, as a sort of the other side of the coin, a possible new molecular mechanism of decline/loss of function leading to a deficit of thyroid hormones formation.


Asunto(s)
Antioxidantes/metabolismo , Diferenciación Celular , Estrés del Retículo Endoplásmico , Mesodermo/citología , Tiroglobulina/metabolismo , Células Epiteliales Tiroideas/citología , Respuesta de Proteína Desplegada , Animales , Células Cultivadas , Regulación de la Expresión Génica , Mesodermo/metabolismo , Ratas , Células Epiteliales Tiroideas/metabolismo
5.
Biochim Biophys Acta ; 1863(6 Pt A): 1146-56, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26940722

RESUMEN

Adipocyte differentiation is critical in obesity. By controlling new adipocyte recruitment, adipogenesis contrasts adipocyte hypertrophy and its adverse consequences, such as insulin resistance. Contrasting data are present in literature on the effect of endoplasmic reticulum (ER) stress and subsequent unfolded protein response (UPR) on adipocyte differentiation, being reported to be either necessary or inhibitory. In this study, we sought to clarify the effect of ER stress and UPR on adipocyte differentiation. We have used two different cell lines, the widely used pre-adipocyte 3T3-L1 cells and a murine multipotent mesenchymal cell line, W20-17 cells. A strong ER stress activator, thapsigargin, and a pathologically relevant inducer of ER stress, glucosamine (GlcN), induced ER stress and UPR above those occurring in the absence of perturbation and inhibited adipocyte differentiation. Very low concentrations of 4-phenyl butyric acid (PBA, a chemical chaperone) inhibited only the overactivation of ER stress and UPR elicited by GlcN, leaving unaltered the part physiologically activated during differentiation, and reversed the inhibitory effect of GlcN on differentiation. In addition, GlcN stimulated proinflammatory cytokine release and PBA prevented these effects. An inhibitor of NF-kB also reversed the effects of GlcN on cytokine release. These results indicate that while ER stress and UPR activation is "physiologically" activated during adipocyte differentiation, the "pathologic" part of ER stress activation, secondary to a glucotoxic insult, inhibits differentiation. In addition, such a metabolic insult, causes a shift of the preadipocyte/adipocyte population towards a proinflammatory phenotype.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular/fisiología , Citocinas/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Mediadores de Inflamación/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adulto , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Citocinas/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Expresión Génica/efectos de los fármacos , Glucosamina/farmacología , Humanos , Ratones , Persona de Mediana Edad , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Fenotipo , Fenilbutiratos/farmacología , Fenilendiaminas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tapsigargina/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
6.
Endocr Rev ; 37(1): 2-36, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26595189

RESUMEN

Thyroglobulin (Tg) is a vertebrate secretory protein synthesized in the thyrocyte endoplasmic reticulum (ER), where it acquires N-linked glycosylation and conformational maturation (including formation of many disulfide bonds), leading to homodimerization. Its primary functions include iodide storage and thyroid hormonogenesis. Tg consists largely of repeating domains, and many tyrosyl residues in these domains become iodinated to form monoiodo- and diiodotyrosine, whereas only a small portion of Tg structure is dedicated to hormone formation. Interestingly, evolutionary ancestors, dependent upon thyroid hormone for development, synthesize thyroid hormones without the complete Tg protein architecture. Nevertheless, in all vertebrates, Tg follows a strict pattern of region I, II-III, and the cholinesterase-like (ChEL) domain. In vertebrates, Tg first undergoes intracellular transport through the secretory pathway, which requires the assistance of thyrocyte ER chaperones and oxidoreductases, as well as coordination of distinct regions of Tg, to achieve a native conformation. Curiously, regions II-III and ChEL behave as fully independent folding units that could function as successful secretory proteins by themselves. However, the large Tg region I (bearing the primary T4-forming site) is incompetent by itself for intracellular transport, requiring the downstream regions II-III and ChEL to complete its folding. A combination of nonsense mutations, frameshift mutations, splice site mutations, and missense mutations in Tg occurs spontaneously to cause congenital hypothyroidism and thyroidal ER stress. These Tg mutants are unable to achieve a native conformation within the ER, interfering with the efficiency of Tg maturation and export to the thyroid follicle lumen for iodide storage and hormonogenesis.


Asunto(s)
Retículo Endoplásmico/fisiología , Modelos Biológicos , Modelos Moleculares , Tiroglobulina/fisiología , Glándula Tiroides/fisiología , Animales , Hipotiroidismo Congénito/genética , Hipotiroidismo Congénito/metabolismo , Hipotiroidismo Congénito/fisiopatología , Estrés del Retículo Endoplásmico , Predisposición Genética a la Enfermedad , Halogenación , Humanos , Mutación , Conformación Proteica , Tiroglobulina/química , Tiroglobulina/genética , Tiroglobulina/metabolismo , Glándula Tiroides/metabolismo , Glándula Tiroides/fisiopatología
7.
PLoS One ; 9(12): e113655, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25489735

RESUMEN

The small scaffold protein PED/PEA-15 is involved in several different physiologic and pathologic processes, such as cell proliferation and survival, diabetes and cancer. PED/PEA-15 exerts an anti-apoptotic function due to its ability to interfere with both extrinsic and intrinsic apoptotic pathways in different cell types. Recent evidence shows that mice overexpressing PED/PEA-15 present larger pancreatic islets and increased beta-cells mass. In the present work we investigated PED/PEA-15 role in hydrogen peroxide-induced apoptosis in Ins-1E beta-cells. In pancreatic islets isolated from Tg(PED/PEA-15) mice hydrogen peroxide-induced DNA fragmentation was lower compared to WT islets. TUNEL analysis showed that PED/PEA-15 overexpression increases the viability of Ins-1E beta-cells and enhances their resistance to apoptosis induced by hydrogen peroxide exposure. The activity of caspase-3 and the cleavage of PARP-1 were markedly reduced in Ins-1E cells overexpressing PED/PEA-15 (Ins-1E(PED/PEA-15)). In parallel, we observed a decrease of the mRNA levels of pro-apoptotic genes Bcl-xS and Bad. In contrast, the expression of the anti-apoptotic gene Bcl-xL was enhanced. Accordingly, DNA fragmentation was higher in control cells compared to Ins-1E(PED/PEA-15) cells. Interestingly, the preincubation with propranolol, an inhibitor of the pathway of PLD-1, a known interactor of PED/PEA-15, responsible for its deleterious effects on glucose tolerance, abolishes the antiapoptotic effects of PED/PEA-15 overexpression in Ins-1E beta-cells. The same results have been obtained by inhibiting PED/PEA-15 interaction with PLD-1 in Ins-1E(PED/PEA-15). These results show that PED/PEA-15 overexpression is sufficient to block hydrogen peroxide-induced apoptosis in Ins-1E cells through a PLD-1 mediated mechanism.


Asunto(s)
Apoptosis/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfolipasa D/metabolismo , Fosfoproteínas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Femenino , Células HeLa , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratas
8.
J Biol Chem ; 289(16): 11488-11496, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24599957

RESUMEN

Newly synthesized thyroglobulin (Tg), the thyroid prohormone, forms detectable high molecular weight mixed disulfide adducts: until now, only Tg "adduct B" was identified as primarily engaging the endoplasmic reticulum oxidoreductases ERp57 and protein disulfide isomerase. Here, we demonstrate that the faster migrating Tg adduct C primarily engages the CaBP1/P5 oxidoreductase, whereas the slower migrating Tg adduct A primarily engages ERp72. Upon siRNA-mediated knockdown of CaBP1/P5 or ERp72, adducts C or A, respectively, are decreased. Within the three Tg adduct bands that do not exhibit a precursor-product relationship, Tg exhibits distinct oxidation patterns. We present evidence suggesting that disulfide maturation occurs within Tg monomers engaged in each of the adduct bands. Moreover, the same Tg substrate molecules can form simultaneous mixed disulfides with both CaBP1/P5 and protein disulfide isomerase, although these are generally viewed as components of distinct oxidoreductase-chaperone protein complexes. Such substrate-oxidoreductase combinations offer Tg the potential for simultaneous oxidative maturation along different parallel tracks leading to the native state.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Tiroglobulina/biosíntesis , Proteínas de Unión al Calcio/genética , Línea Celular , Disulfuros/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Complejos Multiproteicos/genética , Proteína Disulfuro Isomerasas/genética , Tiroglobulina/genética
9.
J Cell Physiol ; 229(10): 1417-26, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24526410

RESUMEN

Recent studies have indicated that endoplasmic reticulum stress, the unfolded protein response activation and altered GRP78 expression can play an important role in a variety of tumors development and progression. Very recently we reported for the first time that GRP78 is increased in endometrial tumors. However, whether GRP78 could play a role in the growth and/or invasiveness of endometrial cancer cells is still unknown. Here we report that the silencing of GRP78 expression affects both cell growth and invasiveness of Ishikawa and AN3CA cells, analyzed by the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) and transwell migration assay, respectively. At variance with Ishikawa cells, AN3CA cells showed, besides an endoplasmic reticulum, also a plasma membrane GRP78 localization, evidenced by both immunofluorescence and cell membrane biotinylation experiments. Intriguingly, flow cytometry experiments showed that the treatment with a specific antibody targeting GRP78 C-terminal domain caused apoptosis in AN3CA but not in Ishikawa cells. Induction of apoptosis in AN3CA cells was not mediated by the p53 pathway activation but was rather associated to reduced AKT phosphorylation. Interestingly, immunofluorescence analysis evidenced that endometrioid adenocarcinoma tissues displayed, similarly to AN3CA cells, also a GRP78 plasma membrane localization. These data suggest that GRP78 and its plasma membrane localization, might play a role in endometrial cancer development and progression and might constitute a novel target for the treatment of endometrial cancer.


Asunto(s)
Carcinoma Endometrioide/metabolismo , Movimiento Celular , Proliferación Celular , Neoplasias Endometriales/metabolismo , Proteínas de Choque Térmico/metabolismo , Apoptosis , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patología , Línea Celular Tumoral , Membrana Celular/metabolismo , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Chaperón BiP del Retículo Endoplásmico , Femenino , Proteínas de Choque Térmico/genética , Humanos , Invasividad Neoplásica , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/metabolismo
10.
J Appl Toxicol ; 33(6): 451-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22120598

RESUMEN

Resin-based dental restorative materials release residual monomers that may affect the vitality of pulp cells. The purpose of this study was to evaluate the cytotoxic effect of two light-cured restorative materials with and without bis-GMA resin, respectively (Clearfil Majesty Posterior and Clearfil Majesty Flow) and a self-curing one (Clearfil DC Core Automix) when applied to the fibroblast cell line NIH-3T3. Samples of the materials were light-cured and placed directly in contact to cells for 24, 48, 72 and 96 h. Cytotoxicity was evaluated by measuring cell death by flow cytometry, cell proliferation by proliferation curves analysis and morphological changes by optical microscopy analysis. All the composite materials tested caused a decrease in cell proliferation, albeit at different degrees. However, only Clearfil DC Core Automix induced cell death, very likely by increasing apoptosis. Morphological alteration of treated cells was also evident, particularly in the Clearfil DC Core Automix-treated cells. The different cytotoxic effects of dental composites should be considered when selecting an appropriate resin-based dental restorative material for operative restorations.


Asunto(s)
Resinas Compuestas/toxicidad , Resinas Sintéticas/toxicidad , Células 3T3 , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colorantes , Tratamiento Restaurativo Atraumático Dental , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Citometría de Flujo , Cinética , Ratones , Necrosis/patología , Propidio
11.
Gynecol Oncol ; 125(1): 220-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22146569

RESUMEN

OBJECTIVES: Endometrial cancer is the most common malignancy of the female genital tract. However, in spite of a huge advance in our understanding of endometrial cancer biology, therapeutic modalities haven't significantly changed over the past 40 years. The activation of the Unfolded Protein Response (UPR) and GRP78 increase following Endoplasmic Reticulum (ER) stress have been recently identified as mechanisms favoring growth, invasion and resistance to therapy of different types of cancer. However, a possible role of ER stress and GRP78 in endometrial cancer has never been investigated. METHODS: Tissue specimens from normal and neoplastic endometrium were analyzed for the expression of the ER stress markers GRP78, ATF6 and CHOP by Real-Time RT-PCR. In addition, GRP78 protein expression and localization were evaluated by Western blot and immunohistochemistry, respectively. The effect of GRP78 knock down on cell growth of Ishikawa cells was analyzed by proliferation curve analysis. RESULTS: In this analysis, the expression levels of GRP78, ATF6 and CHOP mRNAs were significantly increased in specimens of endometrioid endometrial carcinomas. GRP78 and ATF6 protein expression levels were also increased in specimens of endometrial adenocarcinomas. GRP78 knock down caused a decrease of Ishikawa cells' growth. CONCLUSIONS: The increased expression of ER stress markers in endometrioid endometrial carcinomas suggests a role for ER stress, the UPR and, possibly, GRP78 in endometrial cancer. Whether these mechanisms have a substantial function in the pathogenesis of malignant transformation of human endometrium is still under investigation in our laboratory.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Adenocarcinoma/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Endometriales/metabolismo , Estrés del Retículo Endoplásmico , Proteínas de Choque Térmico/metabolismo , Factor de Transcripción CHOP/metabolismo , Western Blotting , Carcinoma Endometrioide/metabolismo , Línea Celular Tumoral , Proliferación Celular , Chaperón BiP del Retículo Endoplásmico , Femenino , Humanos , Inmunohistoquímica , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Respuesta de Proteína Desplegada
12.
J Biol Chem ; 286(38): 33045-52, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21816825

RESUMEN

In vertebrates, the thyroglobulin (Tg) gene product must be exported to the lumen of thyroid follicles for thyroid hormone synthesis. In toto, Tg is composed of multiple type-1 repeats connected by linker and hinge (altogether considered as "region I," nearly 1,200 residues); regions II-III (~720 residues); and cholinesterase-like (ChEL) domain (~570 residues). Regions II-III and ChEL rapidly acquire competence for secretion, yet regions I-II-III require 20 min to become a partially mature disulfide isomer; stabilization of a fully oxidized form requires ChEL. Transition from partially mature to mature Tg occurs as a discrete "jump" in mobility by nonreducing SDS-PAGE, suggesting formation of at most a few final pairings of Cys residues that may be separated by significant intervening primary sequence. Using two independent approaches, we have investigated which portion of Tg is engaged in this late stage of its maturation. First, we demonstrate that this event is linked to oxidation involving region I. Introduction of the Tg-C1245R mutation in the hinge (identical to that causing human goitrous hypothyroidism) inhibits this maturation, although the Cys-1245 partner remains unidentified. Second, we find that Tg truncated after its fourth type-1 repeat is a fully independent secretory protein. Together, the data indicate that final acquisition of secretory competence includes conformational maturation in the interval between linker and hinge segments of region I.


Asunto(s)
Procesamiento Proteico-Postraduccional , Tiroglobulina/química , Tiroglobulina/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Proteínas Mutantes/metabolismo , Oxidación-Reducción , Mutación Puntual/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Secuencias Repetitivas de Aminoácido
13.
Neoplasia ; 13(2): 120-30, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21403838

RESUMEN

Activation of the Ras-Raf-extracellular signal-regulated kinase (ERK) pathway causes not only proliferation and suppression of apoptosis but also the antioncogenic response of senescence. How these contrasting effects are reconciled to achieve cell transformation and cancer formation is poorly understood. In a system of two-step carcinogenesis (dedifferentiated PC EIA, transformed PC EIA-polyoma-middle T [PC EIA + Py] and PC EIA-v-raf [PC EIA + raf] cells], v-raf cooperated with EIA by virtue of a strong prosurvival effect, not elicited by Py-middle T, evident toward serum-deprivation-and H(2)O(2)-induced apoptosis. Apoptosis was detected by DNA fragmentation and annexin V staining. The prosurvival function of v-raf was, in part, mitogen-activated protein kinase/ERK kinase (MEK)-dependent, as shown by pharmacological MEK inhibition. The MEK-dependent antiapoptotic effect of v-raf was exerted despite a lower level of P-ERK1/2 in EIA + raf cells with respect to EIA + Py/EIA cells, which was dependent on a high tyrosine phosphatase activity, as shown by orthovanadate blockade. An ERK1/2 tyrosine phosphatase was likely involved. The high tyrosine phosphatase activity was instrumental to the complete suppression of senescence, detected by ß-galactosidase activity, because tyrosine phosphatase blockade induced senescence in EIA + raf but not in EIA + Py cells. High tyrosine phosphatase activity and evasion from senescence were confirmed in an anaplastic thyroid cancer cell line. Therefore, besides EIA, EIA + raf cells suppress senescence through a new mechanism, namely, phosphatase-mediated P-ERK1/2 inhibition, but, paradoxically, retain the oncogenic effects of the Raf-ERK pathway. We propose that the survival effect of Raf is not a function of absolute P-ERK1/2 levels at a given time but is rather dynamically dependent on greater variations after an apoptotic stimulus.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Oncogénicas v-raf/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Neoplasias de la Tiroides/enzimología , Apoptosis/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Senescencia Celular , Regulación de la Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Proteínas Oncogénicas v-raf/genética , beta-Galactosidasa/metabolismo
14.
J Biol Chem ; 285(23): 17564-73, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20353937

RESUMEN

Thyroglobulin (Tg, precursor for thyroid hormone synthesis) is a large secreted glycoprotein composed of upstream regions I-II-III, followed by the approximately 570 residue cholinesterase-like (ChEL) domain. ChEL has two identified functions: 1) homodimerization, and 2) binding to I-II-III that facilitates I-II-III oxidative maturation required for intracellular protein transport. Like its homologs in the acetylcholinesterase (AChE) family, ChEL possesses two carboxyl-terminal alpha-helices. We find that a Tg-AChE chimera (swapping AChE in place of ChEL) allows for dimerization with monomeric AChE, proving exposure of the carboxyl-terminal helices within the larger context of Tg. Further, we establish that perturbing trans-helical interaction blocks homodimerization of the Tg ChEL domain. Additionally, ChEL can associate with neuroligins (a related family of cholinesterase-like proteins), demonstrating potential for Tg cross-dimerization between non-identical partners. Indeed, when mutant rdw-Tg (Tg-G2298R, defective for protein secretion) is co-expressed with wild-type Tg, the two proteins cross-dimerize and secretion of rdw-Tg is partially restored. Moreover, we find that AChE and soluble neuroligins also can bind to the upstream Tg regions I-II-III; however, they cannot rescue secretion, because they cannot facilitate oxidative maturation of I-II-III. These data suggest that specific properties of distinct Tg ChEL mutants may result in distinct patterns of Tg monomer folding, cross-dimerization with wild-type Tg, and variable secretion behavior in heterozygous patients.


Asunto(s)
Colinesterasas/química , Tiroglobulina/química , Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/química , Dimerización , Heterocigoto , Humanos , Ratones , Chaperonas Moleculares/química , Mutación , Estrés Oxidativo , Pliegue de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química
15.
J Biol Chem ; 284(19): 12752-61, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19276074

RESUMEN

The carboxyl-terminal cholinesterase-like (ChEL) domain of thyroglobulin (Tg) has been identified as critically important in Tg export from the endoplasmic reticulum. In a number of human kindreds suffering from congenital hypothyroidism, and in the cog congenital goiter mouse and rdw rat dwarf models, thyroid hormone synthesis is inhibited because of mutations in the ChEL domain that block protein export from the endoplasmic reticulum. We hypothesize that Tg forms homodimers through noncovalent interactions involving two predicted alpha-helices in each ChEL domain that are homologous to the dimerization helices of acetylcholinesterase. This has been explored through selective epitope tagging of dimerization partners and by inserting an extra, unpaired Cys residue to create an opportunity for intermolecular disulfide pairing. We show that the ChEL domain is necessary and sufficient for Tg dimerization; specifically, the isolated ChEL domain can dimerize with full-length Tg or with itself. Insertion of an N-linked glycan into the putative upstream dimerization helix inhibits homodimerization of the isolated ChEL domain. However, interestingly, co-expression of upstream Tg domains, either in cis or in trans, overrides the dimerization defect of such a mutant. Thus, although the ChEL domain provides a nidus for Tg dimerization, interactions of upstream Tg regions with the ChEL domain actively stabilizes the Tg dimer complex for intracellular transport.


Asunto(s)
Colinesterasas/química , Tiroglobulina/metabolismo , Hormonas Tiroideas/biosíntesis , Secuencia de Aminoácidos , Animales , Células Cultivadas , Epítopos , Glicosilación , Proteínas Fluorescentes Verdes/metabolismo , Inmunoprecipitación , Riñón/citología , Riñón/metabolismo , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/metabolismo , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Homología de Secuencia de Aminoácido , Tiroglobulina/química , Tiroglobulina/genética
16.
J Clin Invest ; 118(8): 2950-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18596923

RESUMEN

Thyroid hormonogenesis requires secretion of thyroglobulin, a protein comprising Cys-rich regions I, II, and III (referred to collectively as region I-II-III) followed by a cholinesterase-like (ChEL) domain. Secretion of mature thyroglobulin requires extensive folding and glycosylation in the ER. Multiple reports have linked mutations in the ChEL domain to congenital hypothyroidism in humans and rodents; these mutations block thyroglobulin from exiting the ER and induce ER stress. We report that, in a cell-based system, mutations in the ChEL domain impaired folding of thyroglobulin region I-II-III. Truncated thyroglobulin devoid of the ChEL domain was incompetent for cellular export; however, a recombinant ChEL protein ("secretory ChEL") was secreted efficiently. Coexpression of secretory ChEL with truncated thyroglobulin increased intracellular folding, promoted oxidative maturation, and facilitated secretion of region I-II-III, indicating that the ChEL domain may function as an intramolecular chaperone. Additionally, we found that the I-II-III peptide was cosecreted and physically associated with secretory ChEL. A functional ChEL domain engineered to be retained intracellularly triggered oxidative maturation of I-II-III but coretained I-II-III, indicating that the ChEL domain may also function as a molecular escort. These insights into the role of the ChEL domain may represent potential therapeutic targets in the treatment of congenital hypothyroidism.


Asunto(s)
Chaperonas Moleculares/metabolismo , Tiroglobulina/química , Línea Celular , Colinesterasas/química , Retículo Endoplásmico/metabolismo , Humanos , Riñón/citología , Mutagénesis Sitio-Dirigida , Mutación , Oxidación-Reducción , Pliegue de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tiroglobulina/genética , Tiroglobulina/metabolismo , Transfección
17.
J Cell Sci ; 121(Pt 4): 477-86, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18211961

RESUMEN

Conditions perturbing the homeostasis of the endoplasmic reticulum (ER) cause accumulation of unfolded proteins and trigger ER stress. In PC Cl3 thyroid cells, thapsigargin and tunicamycin interfered with the folding of thyroglobulin, causing accumulation of this very large secretory glycoprotein in the ER. Consequently, mRNAs encoding BiP and XBP-1 were induced and spliced, respectively. In the absence of apoptosis, differentiation of PC Cl3 cells was inhibited. mRNA and protein levels of the thyroid-specific genes encoding thyroglobulin, thyroperoxidase and the sodium/iodide symporter and of the genes encoding the thyroid transcription factors TTF-1, TTF-2 and Pax-8 were dramatically downregulated. These effects were, at least in part, transcriptional. Moreover, they were selective and temporally distinct from the general and transient PERK-dependent translational inhibition. Thyroid dedifferentiation was accompanied by changes in the organization of the polarized epithelial monolayer. Downregulation of the mRNA encoding E-cadherin, and upregulation of the mRNAs encoding vimentin, alpha-smooth muscle actin, alpha(1)(I) collagen and SNAI1/SIP1, together with formation of actin stress fibers and loss of trans-epithelial resistance were found, confirming an epithelial-mesenchymal transition (EMT). The thyroid-specific and epithelial dedifferentiation by thapsigargin or tunicamycin were completely prevented by the PP2 inhibitor of Src-family kinases and by stable expression of a dominant-negative Src. Together, these data indicate that ER stress induces dedifferentiation and an EMT-like phenotype in thyroid cells through a Src-mediated signaling pathway.


Asunto(s)
Diferenciación Celular/fisiología , Retículo Endoplásmico/metabolismo , Células Epiteliales/citología , Mesodermo/citología , Glándula Tiroides/citología , Animales , Northern Blotting , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Tapsigargina/farmacología , Tiroglobulina/genética , Tiroglobulina/metabolismo , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Tunicamicina/farmacología
18.
J Endocrinol ; 190(3): 641-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17003265

RESUMEN

In PC Cl3 cells, a continuous, fully differentiated rat thyroid cell line, P2Y(2) purinoceptor activation provoked a transient increase of [Ca(2+)](i), followed by a decreasing sustained phase. The alpha and beta1 protein kinase C (PKC) inhibitor Gö6976 decreased the rate of decrement to the basal [Ca(2+)](i) level and increased the peak of Ca(2+) entry of the P2Y(2)-provoked Ca(2+)transients. These effects of Gö 6976 were not caused by an increased permeability of the plasma membrane, since the Mn(2+) and Ba(2+) uptake were not changed by Gö 6976. Similarly, the Na(+)/Ca(2+) exchanger was not implicated, since the rate of decrement to the basal [Ca(2+)](i) level was equally decreased in physiological and Na(+)-free buffers, in the presence of Gö 6976. On the contrary, the activity of the sarcoplasmic-endoplasmic reticulum Ca(2+)ATPase (SERCA) 2b was profoundly affected by Gö 6976 since the drug was able to completely inhibit the stimulation of the SERCA 2b activity elicited by P2-purinergic agonists. Finally, the PKC activator phorbol myristate acetate had effects opposite to Gö 6976, in that it markedly increased the rate of decrement to the basal [Ca(2+)](i) level after P2Y(2) stimulation and also increased the activity of SERCA 2b. These results suggest that SERCA 2b plays a role in regulating the sustained phase of Ca(2+) transients caused by P2Y(2) stimulation.


Asunto(s)
Calcio/metabolismo , Receptores Purinérgicos P2/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Retículo Sarcoplasmático/enzimología , Glándula Tiroides/enzimología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Bario/metabolismo , Transporte Biológico Activo , Calcio/análisis , Carbazoles/farmacología , Línea Celular , Activación Enzimática , Indoles/farmacología , Maleimidas/farmacología , Manganeso/metabolismo , Microscopía Fluorescente , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Receptores Purinérgicos P2Y2 , Acetato de Tetradecanoilforbol/farmacología , Tapsigargina/análisis , Uridina Trifosfato/metabolismo , Uridina Trifosfato/farmacología
19.
Bioelectromagnetics ; 27(7): 560-77, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16724329

RESUMEN

The present work was a comparative study of the bio-effects induced by exposure to 6 mT static magnetic field (MF) on several primary cultures and cell lines. Particular attention was dedicated to apoptosis. Cell viability, proliferation, intracellular Ca(2+) concentration and morphology were also examined. Primary cultures of human lymphocytes, mice thymocytes and cultures of 3DO, U937, HeLa, HepG2 and FRTL-5 cells were grown in the presence of 6 mT static MF and different apoptosis-inducing agents (cycloheximide, H(2)O(2), puromycin, heat shock, etoposide). Biological effects of static MF exposure were found in all the different cells examined. They were cell type-dependent but apoptotic inducer-independent. A common effect of the exposure to static MF was the promotion of apoptosis and mitosis, but not of necrosis or modifications of the cell shape. Increase of the intracellular levels of Ca(2+) ions were also observed. When pro-apoptotic drugs were combined with static MF, the majority of cell types rescued from apoptosis. To the contrary, apoptosis of 3DO cells was significantly increased under simultaneous exposure to static MF and incubation with pro-apoptotic drugs. From these data we conclude that 6 mT static MF exposure interfered with apoptosis in a cell type- and exposure time-dependent manner, while the effects of static MF exposure on the apoptotic program were independent of the drugs used.


Asunto(s)
Apoptosis/efectos de la radiación , Magnetismo/efectos adversos , Animales , Calcio/metabolismo , División Celular/efectos de la radiación , Línea Celular/efectos de la radiación , Células Cultivadas/efectos de la radiación , Células HeLa/efectos de la radiación , Humanos , Hígado/citología , Hígado/efectos de la radiación , Linfocitos/efectos de la radiación , Masculino , Ratones , Timo/citología , Timo/efectos de la radiación , Glándula Tiroides/citología , Glándula Tiroides/efectos de la radiación
20.
Biochem Pharmacol ; 71(1-2): 50-60, 2005 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-16297374

RESUMEN

The effects of cisplatin (cisPt) on the extra cellular signal-regulated kinase (ERK) and the protein kinase B (PKB/Akt), known to play important roles in promoting cell survival and in down regulating apoptosis, were investigated in thyroid cell lines. The cytotoxic effect of cisPt was highest in normal PC-Cl3 cells, intermediate in dedifferentiated PC-E1A and PC-raf cells and lowest in fully transformed and tumorigenic PC-E1Araf cells. CisPt provoked ERK phosphorylation; such phosphorylation was unaltered by Gö6976, a conventional PKC inhibitor, whilst blocked by low doses (0.1 microM) or high doses (10 microM) of GF109203X, an inhibitor of all PKC isozymes, in PC-Cl3 and in PC-E1Araf cells, respectively. In PC-E1Araf, but not in PC-Cl3 cells, the cisPt-provoked ERK phosphorylation was also blocked by a myristoylated PKC-zeta pseudo substrate peptide (PS-zeta). The cytotoxic effects of cisPt increased when cells were pre-incubated with the mitogen-activated protein kinase (MEK) inhibitor PD98059. CisPt provoked the phosphorylation of PKB/Akt and this effect was blocked by LY294002, a PI3K inhibitor. In PC-Cl3 cells pre-incubated with LY294002 the effects of cisPt on ERK phosphorylation and cell mortality resulted unaffected; conversely, LY294002 reduced the ERK phosphorylation and increased cisPt cytotoxity of in PC-E1Araf cells. Furthermore, in PC-E1Araf cells pre-incubated with LY294002 and PS-zeta ERK phosphorylation was abolished and cisPt cytotoxicity was highest. Altogether results highlight a role for PKCs in the upstream regulation of ERK pathway facing the cell response to cisPt treatments. Understanding the mechanisms by which cells process cisPt provides important insights for designing more efficient platinum-based drugs.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Glándula Tiroides/efectos de los fármacos , Animales , Western Blotting , Diferenciación Celular , Línea Celular , Línea Celular Transformada , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Glándula Tiroides/citología , Glándula Tiroides/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA